首页 > 最新文献

Current cancer drug targets最新文献

英文 中文
Anti-Resistant Strategies: Icotinib Derivatives as Promising Non-Small Cell Lung Cancer Therapeutics. 抗逆策略:作为有前途的非小细胞肺癌治疗药物的伊科替尼衍生物。
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-07-04 DOI: 10.2174/0115680096302595240605114828
Zhiwei Zhao, Yu Du, Xiaojie Chen

Background: Non-small cell lung cancer (NSCLC) patients often benefit from EGFR inhibitors like gefitinib. However, drug resistance remains a significant challenge in treatment. The unique properties of 1,2,3-triazole, a nitrogen-based compound, hold promise as potential solutions due to its versatile structural attributes and diverse biological effects, including anticancer properties.

Materials and methods: Our synthesis process involved the huisgen cycloaddition chemical method, which generated diverse icotinib derivatives. We evaluated the anticancer capabilities of these derivatives against various cancer cell lines, with a specific focus on NSCLC cells that exhibit drug resistance. Additionally, we investigated the binding affinity of selected compounds, including 3l, towards wild-type EGFR using surface plasmon resonance (SPR) experiments.

Results: Notably, icotinib derivatives such as derivative 3l demonstrated significant efficacy against different cancer cell lines, including those resistant to conventional therapies. Compound 3l exhibited potent activity with IC50 values below 10 μM against drug-resistant cells. SPR experiments revealed that 3l exhibited enhanced affinity towards wild-type EGFR compared to icotinib. Our research findings suggest that 3l acts as a compelling antagonist for the protein tyrosine kinase of EGFR (EGFR-PTK).

Conclusion: Icotinib derivative 3l, featuring a 1,2,3-triazole ring, demonstrates potent anticancer effects against drug-resistant NSCLC cells. Its enhanced binding affinity to EGFR and modulation of the EGFR-RAS-RAF-MAPK pathway position 3l as a promising candidate for the future development of anticancer drugs.

背景:非小细胞肺癌(NSCLC)患者常常受益于吉非替尼等表皮生长因子受体抑制剂。然而,耐药性仍然是治疗中的一大挑战。1,2,3-三氮唑是一种氮基化合物,由于其独特的结构属性和多种生物效应(包括抗癌特性),有望成为潜在的解决方案:我们的合成过程采用了 Huuisgen 环加成化学方法,生成了多种不同的 icotinib 衍生物。我们评估了这些衍生物对各种癌细胞株的抗癌能力,尤其关注表现出耐药性的 NSCLC 细胞。此外,我们还利用表面等离子体共振(SPR)实验研究了包括 3l 在内的部分化合物与野生型表皮生长因子受体的结合亲和力:结果:值得注意的是,伊柯替尼衍生物(如衍生物 3l)对不同的癌细胞系(包括对传统疗法耐药的癌细胞系)具有显著疗效。化合物 3l 具有强效活性,对耐药细胞的 IC50 值低于 10 μM。SPR 实验显示,与伊柯替尼相比,3l 对野生型表皮生长因子受体的亲和力更强。我们的研究结果表明,3l 是表皮生长因子受体蛋白酪氨酸激酶(表皮生长因子受体-表皮生长因子受体-酪氨酸激酶)的一种令人信服的拮抗剂:结论:具有1,2,3-三唑环的伊柯替尼衍生物3l对耐药NSCLC细胞具有强效抗癌作用。3l与表皮生长因子受体的结合亲和力增强,并能调节表皮生长因子受体-RAS-RAF-MAPK通路,因此有望成为未来开发抗癌药物的候选药物。
{"title":"Anti-Resistant Strategies: Icotinib Derivatives as Promising Non-Small Cell Lung Cancer Therapeutics.","authors":"Zhiwei Zhao, Yu Du, Xiaojie Chen","doi":"10.2174/0115680096302595240605114828","DOIUrl":"https://doi.org/10.2174/0115680096302595240605114828","url":null,"abstract":"<p><strong>Background: </strong>Non-small cell lung cancer (NSCLC) patients often benefit from EGFR inhibitors like gefitinib. However, drug resistance remains a significant challenge in treatment. The unique properties of 1,2,3-triazole, a nitrogen-based compound, hold promise as potential solutions due to its versatile structural attributes and diverse biological effects, including anticancer properties.</p><p><strong>Materials and methods: </strong>Our synthesis process involved the huisgen cycloaddition chemical method, which generated diverse icotinib derivatives. We evaluated the anticancer capabilities of these derivatives against various cancer cell lines, with a specific focus on NSCLC cells that exhibit drug resistance. Additionally, we investigated the binding affinity of selected compounds, including 3l, towards wild-type EGFR using surface plasmon resonance (SPR) experiments.</p><p><strong>Results: </strong>Notably, icotinib derivatives such as derivative 3l demonstrated significant efficacy against different cancer cell lines, including those resistant to conventional therapies. Compound 3l exhibited potent activity with IC50 values below 10 μM against drug-resistant cells. SPR experiments revealed that 3l exhibited enhanced affinity towards wild-type EGFR compared to icotinib. Our research findings suggest that 3l acts as a compelling antagonist for the protein tyrosine kinase of EGFR (EGFR-PTK).</p><p><strong>Conclusion: </strong>Icotinib derivative 3l, featuring a 1,2,3-triazole ring, demonstrates potent anticancer effects against drug-resistant NSCLC cells. Its enhanced binding affinity to EGFR and modulation of the EGFR-RAS-RAF-MAPK pathway position 3l as a promising candidate for the future development of anticancer drugs.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141533893","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Development of CDK12 as a Cancer Therapeutic Target and Related Inhibitors. 将 CDK12 开发为癌症治疗靶点及相关抑制剂。
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-07-04 DOI: 10.2174/0115680096307629240611104728
Biqing Chen, Jiaqi Liu

Cyclin-dependent Kinase 12 (CDK12) is a Cyclin-dependent Kinase (CDK) that plays a crucial role in various biological processes, including transcription, translation, mRNA splicing, cell cycle regulation, and DNA damage repair. Dysregulation of CDK12 has been implicated in tumorigenesis, and genetic alterations affecting CDK12 have been identified in multiple cancer types, including breast cancer, ovarian cancer, gastric cancer, and prostate cancer. Numerous studies have demonstrated that suppression of CDK12 expression effectively inhibits tumor growth and proliferation, underscoring its significance as a cancer biomarker and a potential therapeutic target in cancer treatment. A thorough comprehension of CDK12 is expected to significantly enhance the advancement of novel approaches for the treatment and prevention of cancer. In recent times, endeavors have been undertaken to formulate targeted inhibitors for CDK12, such as PROTAC and molecular gel degraders. Concurrently, investigations have been conducted on the combined utilization of CDK12 small molecule inhibitors and immunotherapy as a potential strategy. This paper examines the diverse functions of CDK12 in the modulation of gene expression and its implications in human tumors. Specifically, it explores the recently uncovered roles of CDK12 kinases in various cellular processes, emphasizing the potential of CDK12 as a viable therapeutic target for the management of human tumors. Furthermore, this review provides an up-to-- date account of the advancements made in utilizing CDK12 in tumor therapy.

细胞周期蛋白依赖性激酶 12(CDK12)是一种细胞周期蛋白依赖性激酶(CDK),在转录、翻译、mRNA 剪接、细胞周期调控和 DNA 损伤修复等各种生物过程中发挥着至关重要的作用。CDK12 的失调与肿瘤发生有关,在多种癌症类型(包括乳腺癌、卵巢癌、胃癌和前列腺癌)中发现了影响 CDK12 的基因改变。大量研究表明,抑制 CDK12 的表达可有效抑制肿瘤的生长和增殖,这凸显了 CDK12 作为癌症生物标志物和癌症治疗潜在靶点的重要意义。对 CDK12 的透彻了解有望大大促进治疗和预防癌症新方法的发展。近来,人们一直在努力研制 CDK12 的靶向抑制剂,如 PROTAC 和分子凝胶降解剂。与此同时,人们还在研究如何将 CDK12 小分子抑制剂和免疫疗法结合起来作为一种潜在的策略。本文探讨了 CDK12 在调控基因表达方面的多种功能及其对人类肿瘤的影响。具体而言,它探讨了最近发现的 CDK12 激酶在各种细胞过程中的作用,强调了 CDK12 作为治疗人类肿瘤的可行靶点的潜力。此外,这篇综述还介绍了在利用 CDK12 治疗肿瘤方面取得的最新进展。
{"title":"Development of CDK12 as a Cancer Therapeutic Target and Related Inhibitors.","authors":"Biqing Chen, Jiaqi Liu","doi":"10.2174/0115680096307629240611104728","DOIUrl":"https://doi.org/10.2174/0115680096307629240611104728","url":null,"abstract":"<p><p>Cyclin-dependent Kinase 12 (CDK12) is a Cyclin-dependent Kinase (CDK) that plays a crucial role in various biological processes, including transcription, translation, mRNA splicing, cell cycle regulation, and DNA damage repair. Dysregulation of CDK12 has been implicated in tumorigenesis, and genetic alterations affecting CDK12 have been identified in multiple cancer types, including breast cancer, ovarian cancer, gastric cancer, and prostate cancer. Numerous studies have demonstrated that suppression of CDK12 expression effectively inhibits tumor growth and proliferation, underscoring its significance as a cancer biomarker and a potential therapeutic target in cancer treatment. A thorough comprehension of CDK12 is expected to significantly enhance the advancement of novel approaches for the treatment and prevention of cancer. In recent times, endeavors have been undertaken to formulate targeted inhibitors for CDK12, such as PROTAC and molecular gel degraders. Concurrently, investigations have been conducted on the combined utilization of CDK12 small molecule inhibitors and immunotherapy as a potential strategy. This paper examines the diverse functions of CDK12 in the modulation of gene expression and its implications in human tumors. Specifically, it explores the recently uncovered roles of CDK12 kinases in various cellular processes, emphasizing the potential of CDK12 as a viable therapeutic target for the management of human tumors. Furthermore, this review provides an up-to-- date account of the advancements made in utilizing CDK12 in tumor therapy.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-07-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141533894","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exosomes Mediate the Production of Oxaliplatin Resistance and Affect Biological Behaviors of Colon Cancer Cell Lines. 外泌体介导奥沙利铂抗药性的产生并影响结肠癌细胞株的生物学行为
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-07-02 DOI: 10.2174/0115680096298783240517050259
Yanwei Ye, Yingze Li, Chu Wu, Yiming Shan, Jie Li, Dongbao Jiang, Jingjing Li, Chao Han, Dongdong Liu, Chunlin Zhao

Background: Colon cancer has high mortality rate which making it one of the leading causes of cancer deaths. Oxaliplatin is a common chemotherapeutic drug, but it has disadvantages such as drug resistance.

Objective: The purpose of this study is to explore the mechanism of exosomes in the resistance of oxaliplatin and verify whether elemene and STAT3 inhibitors reverse the resistance to oxaliplatin.

Methods: Related cell line models were constructed and the proliferation, migration, invasion, apoptosis and resistance to oxaliplatin were evaluated for all three cells of HCT116/L, sensitive cell HCT116 and HCT116+HCT116/L-exosomes (HCT116-exo). It was to explore probable signaling pathways and mechanisms by Western blotting.

Results: HCT116-exo drug-resistant chimeric cells showed greater capacity for proliferation, migration and invasion than HCT116 sensitive cells. After the above cells were treated with oxaliplatin, the apoptosis rate of chimeric drug-resistant cells HCT116-exo and its IC50 increased compared with the sensitive cells HCT116. The proliferation, invasion and migration of cells treated with STAT3 inhibitor or β-elemene combined with oxaliplatin reduced compared with those treated with oxaliplatin or β-elemene alone. The STAT3 inhibitor or β-elemene in combination with oxaliplatin increased the rate of apoptosis relative to oxaliplatin or β-elemene alone. Drug-resistant cell exosomes could promote the EMT process, related to the participation of FGFR4, SHMT2 and STAT3 inhibitors.

Conclusion: Drug-resistant cell exosomes could induce resistance, and improve the capacity of colon cancer towards proliferate, invade, migrate and promote the EMT process. The β-elemene combined with oxaliplatin could reverse the above results which might be related to the STAT3 pathway and EMT pathway in colon cancer.

背景:结肠癌死亡率很高,是导致癌症死亡的主要原因之一。奥沙利铂是一种常见的化疗药物,但它也存在耐药性等缺点:本研究旨在探索外泌体对奥沙利铂耐药的机制,并验证榄香烯和 STAT3 抑制剂是否能逆转奥沙利铂的耐药性:方法:构建相关细胞系模型,评估HCT116/L、敏感细胞HCT116和HCT116+HCT116/L-外泌体(HCT116-exo)三种细胞的增殖、迁移、侵袭、凋亡和对奥沙利铂的耐药性。结果表明:HCT116-exo 细胞中的药物浓度与 HCT116/L 细胞中的 HCT116+HCT116/L- 外泌体浓度呈正相关:结果:与 HCT116 敏感细胞相比,HCT116-exo 耐药嵌合细胞具有更强的增殖、迁移和侵袭能力。用奥沙利铂处理上述细胞后,与敏感细胞HCT116相比,嵌合耐药细胞HCT116-exo的凋亡率及其IC50均有所增加。STAT3抑制剂或β-榄香烯与奥沙利铂联合应用时,细胞的增殖、侵袭和迁移均比单独应用奥沙利铂或β-榄香烯时减少。STAT3抑制剂或β-榄香烯与奥沙利铂联用可提高细胞凋亡率,而奥沙利铂或β-榄香烯单独使用则会降低细胞凋亡率。耐药细胞外泌体可促进EMT过程,这与FGFR4、SHMT2和STAT3抑制剂的参与有关:结论:耐药细胞外泌体可诱导耐药性,并提高结肠癌的增殖、侵袭、迁移和促进EMT过程的能力。β-榄香烯联合奥沙利铂可逆转上述结果,这可能与结肠癌中的 STAT3 通路和 EMT 通路有关。
{"title":"Exosomes Mediate the Production of Oxaliplatin Resistance and Affect Biological Behaviors of Colon Cancer Cell Lines.","authors":"Yanwei Ye, Yingze Li, Chu Wu, Yiming Shan, Jie Li, Dongbao Jiang, Jingjing Li, Chao Han, Dongdong Liu, Chunlin Zhao","doi":"10.2174/0115680096298783240517050259","DOIUrl":"https://doi.org/10.2174/0115680096298783240517050259","url":null,"abstract":"<p><strong>Background: </strong>Colon cancer has high mortality rate which making it one of the leading causes of cancer deaths. Oxaliplatin is a common chemotherapeutic drug, but it has disadvantages such as drug resistance.</p><p><strong>Objective: </strong>The purpose of this study is to explore the mechanism of exosomes in the resistance of oxaliplatin and verify whether elemene and STAT3 inhibitors reverse the resistance to oxaliplatin.</p><p><strong>Methods: </strong>Related cell line models were constructed and the proliferation, migration, invasion, apoptosis and resistance to oxaliplatin were evaluated for all three cells of HCT116/L, sensitive cell HCT116 and HCT116+HCT116/L-exosomes (HCT116-exo). It was to explore probable signaling pathways and mechanisms by Western blotting.</p><p><strong>Results: </strong>HCT116-exo drug-resistant chimeric cells showed greater capacity for proliferation, migration and invasion than HCT116 sensitive cells. After the above cells were treated with oxaliplatin, the apoptosis rate of chimeric drug-resistant cells HCT116-exo and its IC50 increased compared with the sensitive cells HCT116. The proliferation, invasion and migration of cells treated with STAT3 inhibitor or β-elemene combined with oxaliplatin reduced compared with those treated with oxaliplatin or β-elemene alone. The STAT3 inhibitor or β-elemene in combination with oxaliplatin increased the rate of apoptosis relative to oxaliplatin or β-elemene alone. Drug-resistant cell exosomes could promote the EMT process, related to the participation of FGFR4, SHMT2 and STAT3 inhibitors.</p><p><strong>Conclusion: </strong>Drug-resistant cell exosomes could induce resistance, and improve the capacity of colon cancer towards proliferate, invade, migrate and promote the EMT process. The β-elemene combined with oxaliplatin could reverse the above results which might be related to the STAT3 pathway and EMT pathway in colon cancer.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141491173","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Low Expression of SCN4B Predicts Poor Prognosis in Non-Small Cell Lung Cancer. SCN4B的低表达可预测非小细胞肺癌的不良预后
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-07-02 DOI: 10.2174/0115680096293516240607071915
Xia Li, Weiwei Chen, Shu Jiang, Lianlian Zhang, Hua Huang, Yanan Ji, Qinggan Ni, Chunhua Ling

Background: Sodium voltage-gated channel beta subunit 4 (SCN4B) plays a suppressive role in various tumors. However, the role of SCN4B in non-small cell lung cancer (NSCLC) is not yet clear. This study aims to investigate the expression of SCN4B in NSCLC patients and its correlation with prognosis.

Methods: Firstly, the expression of SCN4B in non-small cell lung cancer (NSCLC) was analyzed using The Cancer Genome Atlas (TCGA) database. Then, differential expression genes (DEGs) were identified using R software. Next, DEG enrichment pathways were analyzed using the R package clusterProfiler. Protein-protein interaction networks were revealed through STRING analysis. A heatmap showed significant differential expression of SCN4B. Further analysis included examining SCN4B expression in a pan-cancer context and its correlation with 24 types of immune cells in NSCLC. Subsequently, quantitative real-time polymerase chain reaction (qRT-PCR), Western Blot, immunohistochemistry, and clinical data were used to validate SCN4B expression and prognostic value in NSCLC patients.

Results: SCN4B mRNA expression in non-small cell lung cancer tissues was significantly lower than in adjacent normal tissues (p < 0.001). Clinical correlation analysis confirmed its association with clinical pathology. Gene set enrichment analysis (GSEA) and tumor immune-related analyses indicated that SCN4B is involved in NSCLC-related Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and participates in immune infiltration. qRT-PCR, Western Blot, and immunohistochemistry also confirmed that SCN4B is downregulated in NSCLC patients and is associated with poor prognosis.

Conclusion: SCN4B is downregulated in tumor tissues of NSCLC patients and is associated with a poor prognosis.

背景:钠电压门控通道 beta 亚基 4(SCN4B)在多种肿瘤中发挥抑制作用。然而,SCN4B 在非小细胞肺癌(NSCLC)中的作用尚不明确。本研究旨在探讨SCN4B在NSCLC患者中的表达及其与预后的相关性:首先,利用癌症基因组图谱(TCGA)数据库分析了SCN4B在非小细胞肺癌(NSCLC)中的表达。然后,使用 R 软件鉴定差异表达基因(DEG)。接着,使用 R 软件包 clusterProfiler 分析 DEG 富集途径。通过 STRING 分析揭示了蛋白质-蛋白质相互作用网络。热图显示了 SCN4B 的显著差异表达。进一步的分析包括研究 SCN4B 在泛癌症中的表达及其与 NSCLC 中 24 种免疫细胞的相关性。随后,研究人员利用定量实时聚合酶链反应(qRT-PCR)、Western Blot、免疫组化和临床数据验证了SCN4B在NSCLC患者中的表达和预后价值:结果:SCN4B mRNA在非小细胞肺癌组织中的表达明显低于邻近的正常组织(p < 0.001)。临床相关性分析证实其与临床病理相关。基因组富集分析(GSEA)和肿瘤免疫相关分析表明,SCN4B参与了NSCLC相关的京都基因组百科全书(KEGG)通路,并参与了免疫浸润:结论:SCN4B在NSCLC患者的肿瘤组织中下调,与预后不良有关。
{"title":"Low Expression of SCN4B Predicts Poor Prognosis in Non-Small Cell Lung Cancer.","authors":"Xia Li, Weiwei Chen, Shu Jiang, Lianlian Zhang, Hua Huang, Yanan Ji, Qinggan Ni, Chunhua Ling","doi":"10.2174/0115680096293516240607071915","DOIUrl":"10.2174/0115680096293516240607071915","url":null,"abstract":"<p><strong>Background: </strong>Sodium voltage-gated channel beta subunit 4 (SCN4B) plays a suppressive role in various tumors. However, the role of SCN4B in non-small cell lung cancer (NSCLC) is not yet clear. This study aims to investigate the expression of SCN4B in NSCLC patients and its correlation with prognosis.</p><p><strong>Methods: </strong>Firstly, the expression of SCN4B in non-small cell lung cancer (NSCLC) was analyzed using The Cancer Genome Atlas (TCGA) database. Then, differential expression genes (DEGs) were identified using R software. Next, DEG enrichment pathways were analyzed using the R package clusterProfiler. Protein-protein interaction networks were revealed through STRING analysis. A heatmap showed significant differential expression of SCN4B. Further analysis included examining SCN4B expression in a pan-cancer context and its correlation with 24 types of immune cells in NSCLC. Subsequently, quantitative real-time polymerase chain reaction (qRT-PCR), Western Blot, immunohistochemistry, and clinical data were used to validate SCN4B expression and prognostic value in NSCLC patients.</p><p><strong>Results: </strong>SCN4B mRNA expression in non-small cell lung cancer tissues was significantly lower than in adjacent normal tissues (p < 0.001). Clinical correlation analysis confirmed its association with clinical pathology. Gene set enrichment analysis (GSEA) and tumor immune-related analyses indicated that SCN4B is involved in NSCLC-related Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and participates in immune infiltration. qRT-PCR, Western Blot, and immunohistochemistry also confirmed that SCN4B is downregulated in NSCLC patients and is associated with poor prognosis.</p><p><strong>Conclusion: </strong>SCN4B is downregulated in tumor tissues of NSCLC patients and is associated with a poor prognosis.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141491174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
WITHDRAWN: Use of Tumor-Infiltrating Lymphocyte (TIL) Cell Therapy for Spindle Cell Carcinoma of the Breast: A Case Report 使用肿瘤浸润淋巴细胞 (TIL) 细胞疗法治疗乳腺纺锤形细胞癌:病例报告。
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-07-02 DOI: 10.2174/0115680096294250240604075529
Yiyin Tang, Li Li, Guoqin Wang, Dan Liu, Yinju Yang, Jinyan Yang, Lin Hua, Ruilei Li, Chunlei Ge, Hong Yao, Ke Li

The article has been withdrawn at the request of the authors.

Bentham Science apologizes to the readers of the journal for any inconvenience this may have caused.

The Bentham Editorial Policy on Article Withdrawal can be found at https://benthamscience.com/editorial-policies-main.php

Bentham science disclaimer: It is a condition of publication that manuscripts submitted to this journal have not been published and will not be simul-taneously submitted or published elsewhere. Furthermore, any data, illustration, structure or table that has been pub-lished elsewhere must be reported, and copyright permission for reproduction must be obtained. Plagiarism is strictly forbidden, and by submitting the article for publication the authors agree that the publishers have the legal right to take appropriate action against the authors, if plagiarism or fabricated information is discovered. By submitting a manuscript the authors agree that the copyright of their article is transferred to the publishers if and when the article is accepted for publication.

背景:乳腺纺锤形细胞癌(SCC)是一种罕见的移行细胞癌,具有预后差、复发率高和远处转移等特点。乳腺切除术、化疗、放疗和内分泌治疗是首选的治疗方法。肿瘤浸润淋巴细胞(TIL)疗法是利用患者实体瘤微环境中的免疫细胞来消灭癌细胞,在治疗晚期实体瘤方面取得了良好的效果。然而,将其用于乳腺 SCC 的病例尚未见报道:在此,我们介绍一例将 TIL 治疗与个体化化疗和内分泌治疗相结合治疗乳腺 SCC 的病例。一名 36 岁的中国女性因左侧乳房可触及结节(32 毫米)而就诊。根据乳腺活检和手术标本的组织学和免疫组化分析,她被诊断为乳腺 SCC(IIA 期)。乳房切除术后,患者同时接受了个性化化疗、TIL 治疗和内分泌治疗。她在治疗期间没有出现严重的副作用,随访至少 14 个月后也没有出现局部复发或远处转移:据我们所知,这是第一例证明乳房切除术后 TIL 治疗联合化疗/内分泌治疗对乳腺 SCC 安全有效的病例报告。
{"title":"WITHDRAWN: Use of Tumor-Infiltrating Lymphocyte (TIL) Cell Therapy for Spindle Cell Carcinoma of the Breast: A Case Report","authors":"Yiyin Tang, Li Li, Guoqin Wang, Dan Liu, Yinju Yang, Jinyan Yang, Lin Hua, Ruilei Li, Chunlei Ge, Hong Yao, Ke Li","doi":"10.2174/0115680096294250240604075529","DOIUrl":"10.2174/0115680096294250240604075529","url":null,"abstract":"<p><p>The article has been withdrawn at the request of the authors.</p><p><p>Bentham Science apologizes to the readers of the journal for any inconvenience this may have caused.</p><p><p>The Bentham Editorial Policy on Article Withdrawal can be found at https://benthamscience.com/editorial-policies-main.php</p><p><strong>Bentham science disclaimer: </strong>It is a condition of publication that manuscripts submitted to this journal have not been published and will not be simul-taneously submitted or published elsewhere. Furthermore, any data, illustration, structure or table that has been pub-lished elsewhere must be reported, and copyright permission for reproduction must be obtained. Plagiarism is strictly forbidden, and by submitting the article for publication the authors agree that the publishers have the legal right to take appropriate action against the authors, if plagiarism or fabricated information is discovered. By submitting a manuscript the authors agree that the copyright of their article is transferred to the publishers if and when the article is accepted for publication.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141491175","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Role of Epigenetic Mechanisms in the Treatment of GI Cancers. 表观遗传机制在消化道癌症治疗中的作用。
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-06-28 DOI: 10.2174/0115680096306639240520053736
Ali Shahini, Amirhossein Imani, Amirali Reyhani, Ghazaleh Khalili-Tanha, Elnaz Ghorbani, Hamid Fiuji, Ibrahim Saeed Al-Hayawi, Majid Khazaei, Seyed Mahdi Hassanian, Gordon A Ferns, Elham Nazari, Amir Avan

Epigenetic mechanisms have been shown to play a critical role in the development and progression of gastrointestinal [GI] cancers. These mechanisms involve modifications to DNA and histones that can alter gene expression patterns and may contribute to the initiation and progression of cancers. In recent years, epigenetic therapies have emerged as a promising approach to treating GI cancers. These therapies target specific epigenetic modifications, such as DNA methylation and histone acetylation, to restore normal gene expression patterns and inhibit cancer cell growth. Several epigenetic drugs have been approved for the treatment of GI cancers. Moreover, the use of epigenetic therapies in combination with other treatments, such as chemotherapeutic agents, is being studied to improve treatment outcomes. We have provided an overview of the role of epigenetic mechanisms in GI cancer treatment aimed to focus on recent evidence of the use of epigenetic agents in clinical and preclinical GI cancer studies, including gastric, esophageal, hepatic, pancreatic, and colorectal cancers. Overall, the role of epigenetic mechanisms in GI cancer treatments is an active area of research with the potential to improve patients' treatment outcomes and advance cancer treatment strategies.

表观遗传机制已被证明在胃肠道癌症的发生和发展中起着至关重要的作用。这些机制涉及对 DNA 和组蛋白的修饰,可改变基因表达模式,并可能导致癌症的发生和发展。近年来,表观遗传疗法已成为治疗消化道癌症的一种前景广阔的方法。这些疗法针对特定的表观遗传修饰,如 DNA 甲基化和组蛋白乙酰化,以恢复正常的基因表达模式并抑制癌细胞生长。目前已有几种表观遗传药物被批准用于治疗消化道癌症。此外,目前正在研究将表观遗传疗法与化疗药物等其他疗法结合使用,以改善治疗效果。我们概述了表观遗传机制在消化道癌症治疗中的作用,旨在重点介绍最近在胃癌、食管癌、肝癌、胰腺癌和结直肠癌等消化道癌症临床和临床前研究中使用表观遗传药物的证据。总体而言,表观遗传机制在消化道癌症治疗中的作用是一个活跃的研究领域,有可能改善患者的治疗效果并推进癌症治疗策略。
{"title":"The Role of Epigenetic Mechanisms in the Treatment of GI Cancers.","authors":"Ali Shahini, Amirhossein Imani, Amirali Reyhani, Ghazaleh Khalili-Tanha, Elnaz Ghorbani, Hamid Fiuji, Ibrahim Saeed Al-Hayawi, Majid Khazaei, Seyed Mahdi Hassanian, Gordon A Ferns, Elham Nazari, Amir Avan","doi":"10.2174/0115680096306639240520053736","DOIUrl":"https://doi.org/10.2174/0115680096306639240520053736","url":null,"abstract":"<p><p>Epigenetic mechanisms have been shown to play a critical role in the development and progression of gastrointestinal [GI] cancers. These mechanisms involve modifications to DNA and histones that can alter gene expression patterns and may contribute to the initiation and progression of cancers. In recent years, epigenetic therapies have emerged as a promising approach to treating GI cancers. These therapies target specific epigenetic modifications, such as DNA methylation and histone acetylation, to restore normal gene expression patterns and inhibit cancer cell growth. Several epigenetic drugs have been approved for the treatment of GI cancers. Moreover, the use of epigenetic therapies in combination with other treatments, such as chemotherapeutic agents, is being studied to improve treatment outcomes. We have provided an overview of the role of epigenetic mechanisms in GI cancer treatment aimed to focus on recent evidence of the use of epigenetic agents in clinical and preclinical GI cancer studies, including gastric, esophageal, hepatic, pancreatic, and colorectal cancers. Overall, the role of epigenetic mechanisms in GI cancer treatments is an active area of research with the potential to improve patients' treatment outcomes and advance cancer treatment strategies.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141476150","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Gamma-Tocotrienol Inhibits Proliferation and Growth of HSD17B4-Overexpressing HepG2 Liver Cancer Cells. γ-生育三烯酚抑制 HSD17B4 过度表达的 HepG2 肝癌细胞的增殖和生长
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-06-26 DOI: 10.2174/0115680096319171240623091614
Xiaoming Wang, Xijia Liang, Nan Zhang, Yaqi Wang, Meng Hu, Yun Shi, Min Yao, Lianguo Hou, Lingling Jiang

Introduction: Hydroxysteroid 17-beta dehydrogenase 4 (HSD17B4) is involved in the progression of hepatocellular carcinoma (HCC).

Aims: This study aimed to investigate the inhibitory effect of gamma-tocotrienol (γ-T3) on the proliferation and growth of HSD17B4-overexpressing HepG2 cells.

Methods: HepG2 cells were transfected with empty or HSD17B4-overexpressing plasmids, followed by vitamin E (VE) or γ-T3 treatment. MTS assay, Western blotting, qRT-PCR, and flow cytometry were employed to assess cell proliferation, protein expression, mRNA levels, and apoptosis. HSD17B4 interaction with γ-T3 was assessed by quantifying γ-T3 in the collected precipitate of HSD17B4 using anti-flag magnetic beads. Tumor xenografts were established in NSG mice, and tumor growth was monitored.

Results: HSD17B4 overexpression significantly promoted HepG2 cell proliferation, which was effectively counteracted by VE or γ-T3 treatment in a dose-dependent manner. VE and γ-T3 did not exert their effects through direct regulation of HSD17B4 expression. Instead, γ-T3 was found to interact with HSD17B4, inhibiting its activity in catalyzing the conversion of estradiol (E2) into estrone. Moreover, γ-T3 treatment led to a reduction in cyclin D1 expression and suppressed key proliferation signaling pathways, such as ERK, MEK, AKT, and STAT3. Additionally, γ-T3 promoted apoptosis in HSD17B4-overexpressing HepG2 cells. In an in vivo model, γ-T3 effectively reduced the growth of HepG2 xenograft tumors.

Conclusion: In conclusion, our study demonstrates that γ-T3 exhibits potent anti-proliferative and anti-tumor effects against HepG2 cells overexpressing HSD17B4. These findings highlight the therapeutic potential of γ-T3 in HCC treatment and suggest its role in targeting HSD17B4-associated pathways to inhibit tumor growth and enhance apoptosis.

简介:羟基类固醇17-β脱氢酶4(HSD17B4)参与了肝细胞癌(HCC)的进展:目的:本研究旨在探讨γ-生育三烯酚(γ-T3)对HSD17B4过表达HepG2细胞增殖和生长的抑制作用。方法:用空质粒或 HSD17B4 表达质粒转染 HepG2 细胞,然后处理维生素 E(VE)或γ-T3。采用 MTS 检测、Western 印迹、qRT-PCR 和流式细胞术评估细胞增殖、蛋白表达、mRNA 水平和细胞凋亡。通过使用抗flag磁珠定量检测HSD17B4沉淀物中的γ-T3,评估HSD17B4与γ-T3的相互作用。在NSG小鼠体内建立肿瘤异种移植,并监测肿瘤生长情况:结果:HSD17B4的过表达能显著促进HepG2细胞的增殖,而VE或γ-T3能以剂量依赖的方式有效地抑制HSD17B4的表达。VE 和 γ-T3 并不是通过直接调节 HSD17B4 的表达来发挥作用的。相反,研究发现γ-T3与HSD17B4相互作用,抑制其催化雌二醇(E2)转化为雌酮的活性。此外,γ-T3 还能减少细胞周期蛋白 D1 的表达,抑制关键的增殖信号通路,如 ERK、MEK、AKT 和 STAT3。此外,γ-T3 还能促进过表达 HSD17B4 的 HepG2 细胞凋亡。在体内模型中,γ-T3 能有效减少 HepG2 异种移植肿瘤的生长:总之,我们的研究表明,γ-T3 对过表达 HSD17B4 的 HepG2 细胞具有强效的抗增殖和抗肿瘤作用。这些发现凸显了γ-T3 在治疗 HCC 中的治疗潜力,并表明γ-T3 在靶向 HSD17B4 相关通路以抑制肿瘤生长和增强细胞凋亡方面的作用。
{"title":"Gamma-Tocotrienol Inhibits Proliferation and Growth of HSD17B4-Overexpressing HepG2 Liver Cancer Cells.","authors":"Xiaoming Wang, Xijia Liang, Nan Zhang, Yaqi Wang, Meng Hu, Yun Shi, Min Yao, Lianguo Hou, Lingling Jiang","doi":"10.2174/0115680096319171240623091614","DOIUrl":"https://doi.org/10.2174/0115680096319171240623091614","url":null,"abstract":"<p><strong>Introduction: </strong>Hydroxysteroid 17-beta dehydrogenase 4 (HSD17B4) is involved in the progression of hepatocellular carcinoma (HCC).</p><p><strong>Aims: </strong>This study aimed to investigate the inhibitory effect of gamma-tocotrienol (γ-T3) on the proliferation and growth of HSD17B4-overexpressing HepG2 cells.</p><p><strong>Methods: </strong>HepG2 cells were transfected with empty or HSD17B4-overexpressing plasmids, followed by vitamin E (VE) or γ-T3 treatment. MTS assay, Western blotting, qRT-PCR, and flow cytometry were employed to assess cell proliferation, protein expression, mRNA levels, and apoptosis. HSD17B4 interaction with γ-T3 was assessed by quantifying γ-T3 in the collected precipitate of HSD17B4 using anti-flag magnetic beads. Tumor xenografts were established in NSG mice, and tumor growth was monitored.</p><p><strong>Results: </strong>HSD17B4 overexpression significantly promoted HepG2 cell proliferation, which was effectively counteracted by VE or γ-T3 treatment in a dose-dependent manner. VE and γ-T3 did not exert their effects through direct regulation of HSD17B4 expression. Instead, γ-T3 was found to interact with HSD17B4, inhibiting its activity in catalyzing the conversion of estradiol (E2) into estrone. Moreover, γ-T3 treatment led to a reduction in cyclin D1 expression and suppressed key proliferation signaling pathways, such as ERK, MEK, AKT, and STAT3. Additionally, γ-T3 promoted apoptosis in HSD17B4-overexpressing HepG2 cells. In an in vivo model, γ-T3 effectively reduced the growth of HepG2 xenograft tumors.</p><p><strong>Conclusion: </strong>In conclusion, our study demonstrates that γ-T3 exhibits potent anti-proliferative and anti-tumor effects against HepG2 cells overexpressing HSD17B4. These findings highlight the therapeutic potential of γ-T3 in HCC treatment and suggest its role in targeting HSD17B4-associated pathways to inhibit tumor growth and enhance apoptosis.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141455803","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Decoding the Expressions, Immune Relevance, and Prognostic Values of Ferroptosis Gene TMEM189: A Pan-Cancer Analysis. 解码铁突变基因 TMEM189 的表达、免疫相关性和预后价值:泛癌症分析。
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-06-26 DOI: 10.2174/0115680096308701240605114342
Guanzhong Qiu, Chaoli Song, Meiqing Lou, Jing Lin

Background: TMEM189 is a recently discovered transmembrane protein involved in ether glycerophospholipid synthesis and ferroptosis regulation. However, its role in tumors are not well understood.

Objective: To elucidate the oncogenic effects and prognostic values of TMEM189 in tumors.

Methods: We performed a pan-cancer analysis of TMEM189 using various databases, bioinformatics and statistical tools, and tissue microarray analysis.

Results: TMEM189 is generally upregulated in tumors compared to normal tissues. High TMEM189 expression is linked to reduced promoter methylation. TMEM189 exhibits a negative correlation with immunogenic markers, immune cell infiltration, and expression of immune checkpoint genes (ICGs) in most cancers, implicating its immunosuppressive role in tumor microenvironments (TME). The interacting and similar genes with TMEM189 were involved in hotspot signaling pathways in pan-cancer. TMEM189 overexpression is usually associated with poor prognosis, especially an independent prognostic risk factor for BLCA, BRCA, LUAD, MESO, LIHC and SKCM.

Conclusion: TMEM189 is overexpressed and exerts immunosuppressive effects in many tumors with a significant association with poor prognosis, suggesting its potential as a therapeutic target in cancer treatment.

背景:TMEM189是最近发现的一种跨膜蛋白,参与醚甘油磷脂合成和铁变态调节。然而,人们对其在肿瘤中的作用还不甚了解:目的:阐明 TMEM189 在肿瘤中的致癌作用和预后价值:方法:我们利用各种数据库、生物信息学和统计学工具以及组织芯片分析对 TMEM189 进行了泛癌症分析:结果:与正常组织相比,TMEM189在肿瘤中普遍上调。TMEM189的高表达与启动子甲基化减少有关。在大多数癌症中,TMEM189与免疫原标志物、免疫细胞浸润和免疫检查点基因(ICGs)的表达呈负相关,这表明它在肿瘤微环境(TME)中发挥着免疫抑制作用。与TMEM189相互作用和相似的基因参与了泛癌症的热点信号通路。TMEM189过表达通常与预后不良有关,尤其是BLCA、BRCA、LUAD、MESO、LIHC和SKCM的独立预后风险因素:结论:TMEM189在许多肿瘤中过表达并发挥免疫抑制作用,与预后不良有显著相关性,这表明它有可能成为癌症治疗的靶点。
{"title":"Decoding the Expressions, Immune Relevance, and Prognostic Values of Ferroptosis Gene TMEM189: A Pan-Cancer Analysis.","authors":"Guanzhong Qiu, Chaoli Song, Meiqing Lou, Jing Lin","doi":"10.2174/0115680096308701240605114342","DOIUrl":"https://doi.org/10.2174/0115680096308701240605114342","url":null,"abstract":"<p><strong>Background: </strong>TMEM189 is a recently discovered transmembrane protein involved in ether glycerophospholipid synthesis and ferroptosis regulation. However, its role in tumors are not well understood.</p><p><strong>Objective: </strong>To elucidate the oncogenic effects and prognostic values of TMEM189 in tumors.</p><p><strong>Methods: </strong>We performed a pan-cancer analysis of TMEM189 using various databases, bioinformatics and statistical tools, and tissue microarray analysis.</p><p><strong>Results: </strong>TMEM189 is generally upregulated in tumors compared to normal tissues. High TMEM189 expression is linked to reduced promoter methylation. TMEM189 exhibits a negative correlation with immunogenic markers, immune cell infiltration, and expression of immune checkpoint genes (ICGs) in most cancers, implicating its immunosuppressive role in tumor microenvironments (TME). The interacting and similar genes with TMEM189 were involved in hotspot signaling pathways in pan-cancer. TMEM189 overexpression is usually associated with poor prognosis, especially an independent prognostic risk factor for BLCA, BRCA, LUAD, MESO, LIHC and SKCM.</p><p><strong>Conclusion: </strong>TMEM189 is overexpressed and exerts immunosuppressive effects in many tumors with a significant association with poor prognosis, suggesting its potential as a therapeutic target in cancer treatment.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141455802","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
WEE Family Kinase Inhibitors Combined with Sorafenib Can Selectively Inhibit HCC Cell Proliferation. WEE 家族激酶抑制剂联合索拉非尼可选择性抑制 HCC 细胞增殖
IF 3 4区 医学 Q2 Medicine Pub Date : 2024-06-10 DOI: 10.2174/0115680096298370240520093003
Anling Chen, Ke Yin, Yu Liu, Lei Hu, Qianwen Cui, Xiaofeng Wan, Wulin Yang

Background: Sorafenib is currently the first choice for the treatment of patients with advanced hepatocellular carcinoma, but its therapeutic effect is still limited.

Objectives: This study aims to examine whether WEE family kinase inhibitors can enhance the anticancer effect of sorafenib.

Methods: We analyzed the expression levels of PKMYT1 kinase and WEE1 kinase in HCC, studied the inhibitory effect of PKMYT1 kinase inhibitor RP-6306, WEE1 kinase inhibitor adavosertib combined with sorafenib on the proliferation of HCC cells, and detected the effect of drug combination on CDK1 phosphorylation.

Results: We found that PKMYT1 and WEE1 were upregulated in HCC and were detrimental to patient survival. Cell experiments showed that both RP-6306 and adavosertib (1-100 μM) inhibited the proliferation of HCC cell lines in a dose-dependent manner alone, and the combination of the two drugs had a synergistic effect. In HCC cell lines, sorafenib combined with RP-6306 or adavosertib showed a synergistic antiproliferation effect and less toxicity to normal cells. Sorafenib combined with RP-6306 and adavosertib further inhibited the proliferation of HCC cells and caused complete dephosphorylation of CDK1.

Conclusion: Taken together, our findings provide experimental evidence for the future use of sorafenib in combination with RP-6306 or adavosertib for the treatment of HCC.

背景:索拉非尼是目前治疗晚期肝细胞癌患者的首选药物,但其治疗效果仍然有限:索拉非尼是目前治疗晚期肝细胞癌患者的首选药物,但其治疗效果仍然有限:本研究旨在探讨WEE家族激酶抑制剂能否增强索拉非尼的抗癌效果:我们分析了PKMYT1激酶和WEE1激酶在HCC中的表达水平,研究了PKMYT1激酶抑制剂RP-6306、WEE1激酶抑制剂adavosertib联合索拉非尼对HCC细胞增殖的抑制作用,并检测了联合用药对CDK1磷酸化的影响:结果:我们发现PKMYT1和WEE1在HCC中上调,并对患者的生存不利。细胞实验表明,RP-6306 和 adavosertib(1-100 μM)单独使用对 HCC 细胞株的增殖有剂量依赖性抑制作用,两种药物联合使用有协同作用。在 HCC 细胞系中,索拉非尼与 RP-6306 或 adavosertib 联用具有协同抗增殖作用,对正常细胞的毒性较小。索拉非尼联合 RP-6306 和 adavosertib 可进一步抑制 HCC 细胞的增殖,并导致 CDK1 完全去磷酸化:综上所述,我们的研究结果为今后索拉非尼联合 RP-6306 或 adavosertib 治疗 HCC 提供了实验证据。
{"title":"WEE Family Kinase Inhibitors Combined with Sorafenib Can Selectively Inhibit HCC Cell Proliferation.","authors":"Anling Chen, Ke Yin, Yu Liu, Lei Hu, Qianwen Cui, Xiaofeng Wan, Wulin Yang","doi":"10.2174/0115680096298370240520093003","DOIUrl":"https://doi.org/10.2174/0115680096298370240520093003","url":null,"abstract":"<p><strong>Background: </strong>Sorafenib is currently the first choice for the treatment of patients with advanced hepatocellular carcinoma, but its therapeutic effect is still limited.</p><p><strong>Objectives: </strong>This study aims to examine whether WEE family kinase inhibitors can enhance the anticancer effect of sorafenib.</p><p><strong>Methods: </strong>We analyzed the expression levels of PKMYT1 kinase and WEE1 kinase in HCC, studied the inhibitory effect of PKMYT1 kinase inhibitor RP-6306, WEE1 kinase inhibitor adavosertib combined with sorafenib on the proliferation of HCC cells, and detected the effect of drug combination on CDK1 phosphorylation.</p><p><strong>Results: </strong>We found that PKMYT1 and WEE1 were upregulated in HCC and were detrimental to patient survival. Cell experiments showed that both RP-6306 and adavosertib (1-100 μM) inhibited the proliferation of HCC cell lines in a dose-dependent manner alone, and the combination of the two drugs had a synergistic effect. In HCC cell lines, sorafenib combined with RP-6306 or adavosertib showed a synergistic antiproliferation effect and less toxicity to normal cells. Sorafenib combined with RP-6306 and adavosertib further inhibited the proliferation of HCC cells and caused complete dephosphorylation of CDK1.</p><p><strong>Conclusion: </strong>Taken together, our findings provide experimental evidence for the future use of sorafenib in combination with RP-6306 or adavosertib for the treatment of HCC.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141300213","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Low Expression of GRIM-19 Correlates with Poor Prognosis in Patients with Upper Urinary Tract Urothelial Carcinoma. GRIM-19的低表达与上尿路尿路癌患者的预后不良有关
IF 3 4区 医学 Q2 Medicine Pub Date : 2024-06-05 DOI: 10.2174/0115680096299093240516163839
Feng Tian, Long Lv, Zonglin Liu, Sheng Guan, Fengze Jiang, Qi Wang, Dhan V Kalvakolanu, Sixiong Jiang, Weibing Sun

Purpose: This study aimed to clarify the expression of a gene associated with Retinoid- Interferon-Induced Mortality-19 (GRIM-19) in Upper Urinary Tract Urothelial Carcinoma (UUTUC) and its prognostic significance for UUTUC patients.

Materials and methods: Immunohistochemical (IHC) staining was used to determine the GRIM-19 expression in 70 paired samples. Progression-Free Survival (PFS) and Cancer-Specific Survival (CSS) were assessed using the Kaplan-Meier method. The independent prognostic factors for PFS and CSS were analyzed by multivariable Cox regression models.

Results: IHC staining showed that GRIM-19 expression was significantly decreased in UUTUC, and its cellular location changed from being both cytoplasmic and nuclear to only cytoplasmic. Kaplan- Meier analysis revealed that the patients with tumors expressing low GRIM-19 had a significantly higher risk for tumor progression (P = 0.002) and cancer-specific mortality (P < 0.001) compared to those with high GRIM-19 levels. The Cox regression showed that both GRIM-19 expression (P = 0.025) and lymph node metastasis (LN) (P = 0.007) were independent predictors of progression in the muscle-invasive (MIC) subgroup. GRIM-19 expressions (entire cohort: P = 0.011; MIC subgroup: P = 0.025), LN (entire cohort: P = 0.019; MIC subgroup: P = 0.007), and progression (entire cohort: P < 0.001; MIC subgroup: P < 0.001) were independent predictors of cancer-specific survival.

Conclusion: Low expression of GRIM-19 in patients with UUTUC had significantly shorter PFS or CSS compared to those with high GRIM-19-expressing tumors. High GRIM-19 expression was also strongly associated with longer PFS in MIC patients. It indicates that GRIM-19 might serve as a promising prognostic biomarker for UUTUC patients.

目的:本研究旨在明确视黄醇-干扰素诱导死亡率-19(GRIM-19)相关基因在上尿路尿路上皮癌(UUTUC)中的表达及其对UUTUC患者的预后意义:免疫组化(IHC)染色用于确定70个配对样本中GRIM-19的表达。采用 Kaplan-Meier 法评估无进展生存期(PFS)和癌症特异性生存期(CSS)。通过多变量考克斯回归模型分析了无进展生存期和特异性生存期的独立预后因素:结果:IHC染色显示,GRIM-19在UUTUC中的表达明显下降,其细胞位置也从胞质和细胞核变为仅胞质表达。Kaplan- Meier分析显示,与GRIM-19水平高的患者相比,GRIM-19表达低的患者肿瘤进展风险(P = 0.002)和癌症特异性死亡率(P < 0.001)明显更高。Cox回归显示,GRIM-19表达(P = 0.025)和淋巴结转移(LN)(P = 0.007)是肌肉浸润性(MIC)亚组肿瘤进展的独立预测因素。GRIM-19的表达(整个队列:P = 0.011;MIC亚组:P = 0.025)、淋巴结(整个队列:P = 0.019;MIC亚组:P = 0.007)和病情进展(整个队列:P < 0.001;MIC亚组:P < 0.001)是癌症特异性生存的独立预测因素:结论:与GRIM-19高表达的肿瘤患者相比,GRIM-19低表达的UUTUC患者的PFS或CSS明显较短。GRIM-19的高表达也与MIC患者较长的PFS密切相关。这表明,GRIM-19可作为UUTUC患者的预后生物标志物。
{"title":"Low Expression of GRIM-19 Correlates with Poor Prognosis in Patients with Upper Urinary Tract Urothelial Carcinoma.","authors":"Feng Tian, Long Lv, Zonglin Liu, Sheng Guan, Fengze Jiang, Qi Wang, Dhan V Kalvakolanu, Sixiong Jiang, Weibing Sun","doi":"10.2174/0115680096299093240516163839","DOIUrl":"https://doi.org/10.2174/0115680096299093240516163839","url":null,"abstract":"<p><strong>Purpose: </strong>This study aimed to clarify the expression of a gene associated with Retinoid- Interferon-Induced Mortality-19 (GRIM-19) in Upper Urinary Tract Urothelial Carcinoma (UUTUC) and its prognostic significance for UUTUC patients.</p><p><strong>Materials and methods: </strong>Immunohistochemical (IHC) staining was used to determine the GRIM-19 expression in 70 paired samples. Progression-Free Survival (PFS) and Cancer-Specific Survival (CSS) were assessed using the Kaplan-Meier method. The independent prognostic factors for PFS and CSS were analyzed by multivariable Cox regression models.</p><p><strong>Results: </strong>IHC staining showed that GRIM-19 expression was significantly decreased in UUTUC, and its cellular location changed from being both cytoplasmic and nuclear to only cytoplasmic. Kaplan- Meier analysis revealed that the patients with tumors expressing low GRIM-19 had a significantly higher risk for tumor progression (P = 0.002) and cancer-specific mortality (P < 0.001) compared to those with high GRIM-19 levels. The Cox regression showed that both GRIM-19 expression (P = 0.025) and lymph node metastasis (LN) (P = 0.007) were independent predictors of progression in the muscle-invasive (MIC) subgroup. GRIM-19 expressions (entire cohort: P = 0.011; MIC subgroup: P = 0.025), LN (entire cohort: P = 0.019; MIC subgroup: P = 0.007), and progression (entire cohort: P < 0.001; MIC subgroup: P < 0.001) were independent predictors of cancer-specific survival.</p><p><strong>Conclusion: </strong>Low expression of GRIM-19 in patients with UUTUC had significantly shorter PFS or CSS compared to those with high GRIM-19-expressing tumors. High GRIM-19 expression was also strongly associated with longer PFS in MIC patients. It indicates that GRIM-19 might serve as a promising prognostic biomarker for UUTUC patients.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141283186","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Current cancer drug targets
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1